Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
2.
J Nanobiotechnology ; 22(1): 234, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724978

RESUMEN

Radiotherapy-induced immune activation holds great promise for optimizing cancer treatment efficacy. Here, we describe a clinically used radiosensitizer hafnium oxide (HfO2) that was core coated with a MnO2 shell followed by a glucose oxidase (GOx) doping nanoplatform (HfO2@MnO2@GOx, HMG) to trigger ferroptosis adjuvant effects by glutathione depletion and reactive oxygen species production. This ferroptosis cascade potentiation further sensitized radiotherapy by enhancing DNA damage in 4T1 breast cancer tumor cells. The combination of HMG nanoparticles and radiotherapy effectively activated the damaged DNA and Mn2+-mediated cGAS-STING immune pathway in vitro and in vivo. This process had significant inhibitory effects on cancer progression and initiating an anticancer systemic immune response to prevent distant tumor recurrence and achieve long-lasting tumor suppression of both primary and distant tumors. Furthermore, the as-prepared HMG nanoparticles "turned on" spectral computed tomography (CT)/magnetic resonance dual-modality imaging signals, and demonstrated favorable contrast enhancement capabilities activated by under the GSH tumor microenvironment. This result highlighted the potential of nanoparticles as a theranostic nanoplatform for achieving molecular imaging guided tumor radiotherapy sensitization induced by synergistic immunotherapy.


Asunto(s)
Ferroptosis , Inmunoterapia , Compuestos de Manganeso , Proteínas de la Membrana , Ratones Endogámicos BALB C , Nanopartículas , Nucleotidiltransferasas , Óxidos , Fármacos Sensibilizantes a Radiaciones , Animales , Ratones , Inmunoterapia/métodos , Óxidos/química , Óxidos/farmacología , Femenino , Nucleotidiltransferasas/metabolismo , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Línea Celular Tumoral , Nanopartículas/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/química , Proteínas de la Membrana/metabolismo , Ferroptosis/efectos de los fármacos , Glucosa Oxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Daño del ADN , Microambiente Tumoral/efectos de los fármacos
3.
Aging (Albany NY) ; 16(7): 5866-5886, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38613793

RESUMEN

NXPH4 promotes cancer proliferation and invasion. However, its specific role and mechanism in cancer remain unclear. Transcriptome and clinical data for pan-cancer were derived from the TCGA database. K-M survival curve and univariate Cox were used for prognostic analysis. CIBERSORT and TIMER algorithms were employed to calculate immune cell infiltration. Gene set enrichment analysis (GSEA) was employed for investigating the function of NXPH4. Western blot verified differential expression of NXPH4 in colon cancer. Functional assays (CCK-8, plate clonogenicity assay, wound healing assay, and Transwell assay) confirmed the impact of NXPH4 on proliferation, invasion, and migration of colon cancer cells. Dysregulation of NXPH4 in pan-cancer suggests its potential as a diagnostic and prognostic marker for certain cancers, including colon and liver cancer. High expression of NXPH4 in pan-cancer might be associated with the increase in copy number and hypomethylation. NXPH4 expression in pan-cancer is substantially linked to immune cell infiltration in the immune microenvironment. NXPH4 expression is associated with the susceptibility to immunotherapy and chemotherapy. Western blot further confirmed the higher expression of NXPH4 in colon cancer. Knockdown of NXPH4 significantly suppresses proliferation, invasion, and migration of colon cancer cell lines HT-29 and HCT116, as validated by functional assays.


Asunto(s)
Biomarcadores de Tumor , Movimiento Celular , Proliferación Celular , Neoplasias del Colon , Humanos , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/metabolismo , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Proliferación Celular/genética , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Progresión de la Enfermedad , Células HT29 , Células HCT116 , Pronóstico , Invasividad Neoplásica , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Línea Celular Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
4.
Adv Healthc Mater ; 13(14): e2303626, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38387885

RESUMEN

Immunotherapy has emerged as an innovative strategy with the potential to improve outcomes in cancer patients. Recent evidence indicates that radiation-induced DNA damage can activate the cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway to enhance the antitumor immune response. Even so, only a small fraction of patients currently benefits from radioimmunotherapy due to the radioresistance and the inadequate activation of the cGAS-STING pathway. Herein, this work integrates hafnium oxide (HfO2) nanoparticles (radiosensitizer) and 7-Ethyl-10-hydroxycamptothecin (SN38, chemotherapy drug, STING agonist) into a polydopamine (PDA)-coated core-shell nanoplatform (HfO2@PDA/Fe/SN38) to achieve synergistic chemoradiotherapy and immunotherapy. The co-delivery of HfO2/SN38 greatly enhances radiotherapy efficacy by effectively activating the cGAS-STING pathway, which then triggers dendritic cells maturation and CD8+ T cells recruitment. Consequently, the growth of both primary and abscopal tumors in tumor-bearing mice is efficiently inhibited. Moreover, the HfO2@PDA/Fe/SN38 complexes exhibit favorable magnetic resonance imaging (MRI)/photoacoustic (PA) bimodal molecular imaging properties. In summary, these developed multifunctional complexes have the potential to intensify immune activation to realize simultaneous cancer Radio/Chemo/Immunotherapy for clinical translation.


Asunto(s)
Inmunoterapia , Proteínas de la Membrana , Nanopartículas , Nucleotidiltransferasas , Animales , Nucleotidiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Inmunoterapia/métodos , Nanopartículas/química , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Línea Celular Tumoral , Humanos , Camptotecina/farmacología , Camptotecina/química , Camptotecina/análogos & derivados , Imagen Molecular/métodos , Polímeros/química , Neoplasias/terapia , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Indoles/química , Indoles/farmacología , Femenino
6.
Lab Invest ; 102(11): 1182-1191, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36183046

RESUMEN

Non-small cell lung cancer (NSCLC) is one of the most malignant cancers worldwide. A growing number of studies have suggested that long noncoding RNAs (lncRNAs) play a key role in the progression of non-small cell lung cancer (NSCLC). Here, we report a novel lncRNA DLGAP1 antisense RNA 1 (DLGAP1-AS1) that exhibits oncogenic properties in NSCLC. The lncRNA DLGAP1-AS1 and denticleless protein homolog (DTL) presented upregulated expression, but microRNA-193a-5p (miR-193a-5p) showed downregulated expression in cancerous tissues of human lung samples from 48 patients with NSCLC. Partial loss of lncRNA DLGAP1-AS1 reduced malignant cell viability, migration, and invasion but induced apoptosis. Dual-luciferase reporter gene, RNA pull-down and RNA binding protein immunoprecipitation assays demonstrated enrichment of lncRNA DLGAP1-AS1 in miR-193a-5p and Argonaute 2, suggesting that lncRNA DLGAP1-AS1 modulated DTL, a putative target of miR-193a-5p. We also found that restoration of miR-193a-5p rescued NSCLC cell biological functions affected by overexpression of lncRNA DLGAP1-AS1. Silencing lncRNA DLGAP1-AS1 was found to reduce the tumorigenesis of NSCLC cells xenografted into nude mice, which was rescued by DTL overexpression. In conclusion, our study highlights a novel regulatory network of the lncRNA DLGAP1-AS1/miR-193a-5p/DTL axis in NSCLC, providing a potential therapeutic strategy for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Ratones , Animales , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica , Proliferación Celular/genética , Movimiento Celular/genética , Línea Celular Tumoral , Neoplasias Pulmonares/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Nucleares/genética
7.
Front Bioeng Biotechnol ; 10: 990145, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091467

RESUMEN

Breast cancer (BC) is the most common malignancy among females worldwide, and high resistance to drugs and metastasis rates are the leading causes of death in BC patients. Releasing anti-cancer drugs precisely to the tumor site can improve the efficacy and reduce the side effects on the body. Natural polymers are attracting extensive interest as drug carriers in treating breast cancer. Hyaluronic acid (HA) is a natural polysaccharide with excellent biocompatibility, biodegradability, and non-immunogenicity and is a significant component of the extracellular matrix. The CD44 receptor of HA is overexpressed in breast cancer cells and can be targeted to breast tumors. Therefore, many researchers have developed nano drug delivery systems (NDDS) based on the CD44 receptor tumor-targeting properties of HA. This review examines the application of HA in NDDSs for breast cancer in recent years. Based on the structural composition of NDDSs, they are divided into HA NDDSs, Modified HA NDDSs, and HA hybrid NDDSs.

8.
J Control Release ; 347: 369-378, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35577149

RESUMEN

Chemo-radiotherapy has been extensively used in clinics, displaying substantial advantages in treatment and prognosis. Stimuli-responsive biodegradable nanoagents that can achieve not only delivery and controlled release of chemotherapeutics, but also hypoxia alleviation to enhance chemoradiotherapy therefore has tremendous potential. Herein, glutathione (GSH)-responsive, biodegradable, doxorubicin-carrying hollow mesoporous organotantalum nanospheres modified with Au and Pt dual nanoenzymes (HMOTP@Pt@Au@Dox) were constructed for chemo-radio sensitization. Degradation of HMOTP@Pt@Au@Dox can be self-activated through GSH stimulation and on-demand release packaged Dox owing to the disulfide bond in the hybrid framework of organotantalum nanospheres. Au and Pt nanoenzymes triggered cascade catalytic reactions that could alleviate hypoxia by utilizing ß-d-glucose and H2O2, thereby sensitizing ROS-based chemoradiotherapy with synergistic starving therapy. Given the radiosensitization of high-Z elements (Ta, Pt, Au), nanoenzymes induced cascade catalytic reaction for hypoxia relief, and the depletion of the predominant antioxidant GSH, desirable tumor suppression could be achieved both in vitro and in vivo, indicating that HMOTP@Pt@Au@Dox is a promising nanoagent to boost chemo-radiotherapy.


Asunto(s)
Nanopartículas , Nanosferas , Línea Celular Tumoral , Doxorrubicina , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno , Hipoxia , Nanopartículas/química
9.
Onco Targets Ther ; 15: 571-584, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35615557

RESUMEN

Introduction: From an oncologic perspective, genetic detection is becoming a frontline clinical test, used to identify actionable alterations for targeted therapy, monitor molecular clonal tumor evolution, indicate disease progression and prognosis, and predict medication efficacy and resistance. From analysis of both tumor tissue and cell-free DNA from a large cohort of non-small cell lung cancer patients in East-China, we characterized the full spectrum of genomic alterations. Methods: The study comprised 3000 unpaired samples including 1351 tumor tissue DNA (tDNA) and 1649 cell-free circulating tumor DNA (cfDNA) samples, from which 67 cancer-related genes were sequenced and the genetic alteration profiles were depicted. Integrative molecular analyses identified the frequently mutated genes, uncovered co-occurring somatic alterations, described the distribution of hotspot variants, analyzed the frequency of variant alleles, and notably distinguished actionable, novel variants. Results: The most commonly affected genes were EGFR, TP53, KRAS, CDKN2A, and PIK3CA in both tDNA and cfDNA samples. EGFR and CTNNB1, PIK3CA and PTEN, ERBB2 and SMO were found to have frequent co-occurring alterations in tDNA samples, while EGFR and SMO, KRAS and PDGFRA, PIK3CA and KDR were in cfDNA samples. A large number of primary druggable variants were identified in tDNA samples, while numerous drug-resistance variants, rare actionable variants, and non-EGFR actionable variants were detected in cfDNA samples. Novel variants were enriched in KDR, KIT, TP53, ABL1, FGFR1 in tDNA samples while the majority of novel variants were distributed in PDGFRA, EGFR, KIT, ROS1, BRCA2 in cfDNA samples. Variant allele frequency in tDNA samples was significantly (P < 0.001) higher than that in cfDNA samples. Conclusion: The results revealed considerable differences in the alteration characteristics between the two kinds of specimens. To date, this study represents the largest real-world investigation of its kind, derived from the largest number of patients in East-China. It reinforced and expanded the mechanism of molecular analysis of neoplastic genetic profiles.

10.
Acta Pharm Sin B ; 11(10): 3231-3243, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34729312

RESUMEN

Ferroptosis, as a newly discovered cell death form, has become an attractive target for precision cancer therapy. Several ferroptosis therapy strategies based on nanotechnology have been reported by either increasing intracellular iron levels or by inhibition of glutathione (GSH)-dependent lipid hydroperoxidase glutathione peroxidase 4 (GPX4). However, the strategy by simultaneous iron delivery and GPX4 inhibition has rarely been reported. Herein, novel tumor microenvironments (TME)-activated metal-organic frameworks involving Fe & Cu ions bridged by disulfide bonds with PEGylation (FCSP MOFs) were developed, which would be degraded specifically under the redox TME, simultaneously achieving GSH-depletion induced GPX4 inactivation and releasing Fe ions to produce ROS via Fenton reaction, therefore causing ferroptosis. More ROS could be generated by the acceleration of Fenton reaction due to the released Cu ions and the intrinsic photothermal capability of FCSP MOFs. The overexpressed GSH and H2O2 in TME could ensure the specific TME self-activated therapy. Better tumor therapeutic efficiency could be achieved by doxorubicin (DOX) loading since it can not only cause apoptosis, but also indirectly produce H2O2 to amplify Fenton reaction. Remarkable anti-tumor effect of obtained FCSP@DOX MOFs was verified via both in vitro and in vivo assays.

11.
Bioengineered ; 12(2): 9301-9312, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34723710

RESUMEN

microRNAs (miRNAs), through their regulation of the expression and activity of numerous proteins, are involved in almost all cellular processes. As a consequence, dysregulation of miRNA expression is closely associated with the development and progression of cancers. Recently, DNA methylation has been shown to play a key role in miRNA expression dysregulation in tumors. miRNA-204-5p commonly acts in the suppression of oncogenes in tumors. In this study, the levels of miRNA-204-5p were found to be down-regulated in the astrocytoma samples. miRNA-204-5p expression was also down-regulated in two astrocytoma cell lines (U87MG and LN382). Examination of online databases showed that the miRNA-204-5p promoter regions exist in CpG islands, which might be subjected to differential methylation. Subsequently, we showed that the miRNA-204-5p promoter region was hypermethylated in the astrocytoma tissue samples and cell lines. Then we found that ezrin expression was down-regulated with an increase in miRNA-204-5p expression in LN382 and U87MG cells after 5-aza-2'-deoxycytidine (5'AZA) treatment compared with control DMSO treatment. In addition, LN382 and U87MG cells treated with 5'AZA exhibited significantly inhibited cell invasion and migration . In a recovery experiment, cell invasion and migration returned to normal levels as miRNA-204-5p and ezrin levels were restored. Overall, our study suggests that miRNA-204-5p acts as a tumor suppressor to influence astrocytoma invasion and migration by targeting ezrin and that miRNA-204-5p expression is downregulated by DNA methylation. This study provides a new potential strategy for astrocytoma treatment.


Asunto(s)
Astrocitoma/genética , Movimiento Celular/genética , Proteínas del Citoesqueleto/metabolismo , Metilación de ADN/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , MicroARNs/metabolismo , Astrocitoma/patología , Azacitidina/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , Invasividad Neoplásica
12.
Ann Rheum Dis ; 80(7): 891-902, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33858850

RESUMEN

OBJECTIVES: The aim of this study was to identify the role of tenascin-C (TNC) in entheseal new bone formation and to explore the underlying molecular mechanism. METHODS: Ligament tissue samples were obtained from patients with ankylosing spondylitis (AS) during surgery. Collagen antibody-induced arthritis and DBA/1 models were established to observe entheseal new bone formation. TNC expression was determined by immunohistochemistry staining. Systemic inhibition or genetic ablation of TNC was performed in animal models. Mechanical properties of extracellular matrix (ECM) were measured by atomic force microscopy. Downstream pathway of TNC was analysed by RNA sequencing and confirmed with pharmacological modulation both in vitro and in vivo. Cellular source of TNC was analysed by single-cell RNA sequencing (scRNA-seq) and confirmed by immunofluorescence staining. RESULTS: TNC was aberrantly upregulated in ligament and entheseal tissues from patients with AS and animal models. TNC inhibition significantly suppressed entheseal new bone formation. Functional assays revealed that TNC promoted new bone formation by enhancing chondrogenic differentiation during endochondral ossification. Mechanistically, TNC suppressed the adhesion force of ECM, resulting in the activation of downstream Hippo/yes-associated protein signalling, which in turn increased the expression of chondrogenic genes. scRNA-seq and immunofluorescence staining further revealed that TNC was majorly secreted by fibroblast-specific protein-1 (FSP1)+fibroblasts in the entheseal inflammatory microenvironment. CONCLUSION: Inflammation-induced aberrant expression of TNC by FSP1+fibroblasts promotes entheseal new bone formation by suppressing ECM adhesion forces and activating Hippo signalling.


Asunto(s)
Matriz Extracelular/patología , Osificación Heterotópica/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Espondilitis Anquilosante/metabolismo , Tenascina/metabolismo , Animales , Artritis Experimental , Vía de Señalización Hippo , Humanos , Ratones , Osificación Heterotópica/patología , Transducción de Señal/fisiología , Espondilitis Anquilosante/patología
13.
J Orthop Surg (Hong Kong) ; 29(1): 2309499020988177, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33626976

RESUMEN

PURPOSE: To compare the sagittal alignment of different surgical approaches in patients with multiple levels cervical spondylotic myelopathy and explore the relationship between the cervical sagittal alignment and patient's health relative quality of life. METHOD: A total of 97 multiple levels cervical spondylotic myelopathy patients who underwent surgery from January 2013 to January 2019 were collected in this study. Patients were divided into three groups: anterior cervical discectomy with fusion, anterior cervical corpectomy with fusion and laminectomy with fusion groups. Clinical outcomes and sagittal alignment parameters were compared preoperative and postoperative. RESULTS: There were no significant differences in the average age and sex ratio among the groups. Sagittal parameters correlated to health relative quality of life were C7 slope, occipito-C2 angle, external auditory meatus tilt and cervical sagittal vertical axis. Both anterior cervical discectomy with fusion and anterior cervical corpectomy with fusion groups exhibited better sagittal alignment and clinical outcomes improvement postoperatively. Anterior cervical discectomy with fusion provided better clinical outcomes and the better improvement of cervical lordosis, C7 slope, occipito-C2 angle and cervical sagittal vertical axis compared with patients with Laminectomy with fusion. CONCLUSION: C7 slope, occipito-C2 angle, external auditory meatus tilt and cervical sagittal vertical axis are the most important cervical sagittal parameters correlated to clinical outcomes in patients with multilevels cervical spondylotic myelopathy; anterior cervical discectomy with fusion and anterior cervical corpectomy with fusion provides more efficient to restoration of cervical sagittal alignment.


Asunto(s)
Vértebras Cervicales , Discectomía , Laminectomía , Enfermedades de la Médula Espinal/cirugía , Fusión Vertebral/métodos , Espondilosis/cirugía , Adulto , Anciano , Estudios de Cohortes , Descompresión Quirúrgica/métodos , Femenino , Humanos , Lordosis/complicaciones , Lordosis/cirugía , Masculino , Persona de Mediana Edad , Calidad de Vida , Enfermedades de la Médula Espinal/etiología , Espondilosis/complicaciones , Resultado del Tratamiento
14.
Transl Cancer Res ; 10(9): 4180-4195, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35116714

RESUMEN

BACKGROUND: C-X-C chemokine receptor 4 (CXCR4) is a specific receptor of stromal cell-derived factor-1, also known as CXCL12. The interaction between CXCL12 and its receptor CXCR4 can activate various signaling pathways, including gene expression, cell proliferation, migration, tumorigenesis, angiogenesis, etc. Although there is evidence to support the association between CXCR4 and some cancers, there is no pan-cancer analysis. To fill this gap, we analyzed the role of CXCR4 in cancer-based on The Cancer Genome Atlas (TCGA). METHODS: We used TCGA, Genotype-Tissue Expression (GTEx) and Clinical Proteomic Tumor Analysis Consortium (CPTAC) databases to analyze the expression, variation and phosphorylation of CXCR4 in different cancers. At the same time, we also carried out Kyoto Encyclopedia of Genes (KEGG) and Gene Ontology (GO) enrichment analysis. RESULTS: We found that CXCR4 expression was significantly increased in bladder urothelial carcinoma (BLCA) and other cancers, and CXCR4 expression in BLCA, cervical squamous cell carcinoma (CESC) and other cancers was related to tumor stage. CXCR4 expression was positively correlated with tumor-associated fibroblasts in BLCA, breast adenocarcinoma (BRCA), CESC and other cancers. GO analysis showed that CXCR4-related genes were mainly enriched in biological processes (BPs) and cellular components (CCs). KEGG analysis showed that CXCR4 was mainly involved in "chemokine signaling pathway", "natural killer cell-mediated cytotoxicity", and "JAK-STAT signaling pathway". CONCLUSIONS: The expression of CXCR4 in different cancers has different effects on the prognosis of patients and the infiltration of immune cells.

15.
ACS Appl Mater Interfaces ; 12(5): 5520-5530, 2020 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-31891473

RESUMEN

Radiotherapy (RT) is one of the most widely used cancer treatments in the clinical setting, while hypoxia-associated resistance often occurs. Herein, a PEGylated TaOx-based oxygen-carrying nanoplatform was constructed for triple sensitizing tumor radiotherapy. The high-Z element based hollow mesoporous TaOx nanospheres were prepared following the in situ growth of ultrasmall CuS nanocrystals and then packaged with O2-saturated perfluoropentane (PFP). NIR laser-triggered mild hyperthermia would lead to the increase of intratumoral blood flow, together with the release of O2, the radiotherapeutic efficiency would be enhanced. Alternatively, radiant energy would be deposited inside the tumor by the Ta element, therefore triple sensitization of radiotherapy could be achieved. The in vivo studies showed that the as-prepared nanospheres could achieve almost total inhibition of tumor growth without obvious side effects, which provides new possibilities for multisensitizing tumor radiotherapy.


Asunto(s)
Materiales Biocompatibles/uso terapéutico , Nanosferas/química , Neoplasias/terapia , Óxidos/química , Tantalio/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cobre/química , Femenino , Fluorocarburos/química , Humanos , Hipertermia Inducida , Rayos Infrarrojos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Nanosferas/toxicidad , Neoplasias/patología , Neoplasias/radioterapia , Oxígeno/química , Porosidad , Especies Reactivas de Oxígeno/metabolismo
16.
FASEB J ; 33(9): 10126-10139, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31216173

RESUMEN

Growing evidence shows that the inhibitory effect of inflammatory cytokines on new bone formation by osteogenic precursor cells is a critical cause of net bone-density reduction. Melatonin has been proven to be a potential therapeutic candidate for osteoporosis. However, whether it is capable of antagonizing the suppressing effect of inflammatory cytokines on osteogenic precursor cells is so far elusive. In this study, using the cell culture system of human bone marrow stromal cells and MC3T3-E1 preosteoblasts, we recorded the following vital observations that provided insights of melatonin-induced bone formation: 1) melatonin induced bone formation in both normal and inflammatory conditions; 2) Wnt4 was essential for melatonin-induced bone formation in inflammatory stimulation; 3) melatonin- and Wnt4-induced bone formation occurred via activation of ß-catenin and p38-JNK MAPK pathways by interaction with a distinct frizzled LDL receptor-related protein complex; 4) melatonin suppressed the inhibitory effect of NF-κB on osteogenesis in a Wnt4-dependent manner; and 5) melatonin induced Wnt4 expression through the ERK1/2-Pax2-Egr1 pathway. In summary, we showed a novel mechanism of melatonin-induced bone formation in an inflammatory environment. Melatonin-induced Wnt4 expression is essential for its osteoinductive effect and the inhibitory effect of NF-κB on bone formation. Our novel findings may provide useful information for its potential translational application.-Li, X., Li, Z., Wang, J., Li, Z., Cui, H., Dai, G., Chen, S., Zhang, M., Zheng, Z., Zhan, Z., Liu, H. Wnt4 signaling mediates protective effects of melatonin on new bone formation in an inflammatory environment.


Asunto(s)
Melatonina/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Vía de Señalización Wnt/fisiología , Proteína Wnt4/fisiología , Animales , Calcio/metabolismo , Línea Celular , Receptores Frizzled/fisiología , Regulación de la Expresión Génica , Humanos , Inflamación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , FN-kappa B/fisiología , Osteoblastos/fisiología , Osteogénesis/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Receptores de LDL/fisiología , Receptores Wnt/efectos de los fármacos , Receptores Wnt/fisiología , Factor de Necrosis Tumoral alfa/farmacología
17.
BMC Complement Altern Med ; 17(1): 455, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28886741

RESUMEN

BACKGROUND: Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease, for which there is currently no safe and effective drug for therapy. In this study, we explored the effects of taurine, tea polyphenols (TPs), or a combination thereof, on NASH rats. METHODS: Rats were divided into a normal group, a high-fat diet induced model group and a treatment group (including taurine, TPs, or taurine + TPs treatment for 8 weeks). Twelve weeks later, all rats were sacrificed, and serum transaminase, lipid and lipopolysaccharide levels and hepatic oxidative stress levels were determined. Histological changes were evaluated. RESULTS: In NASH rats, hepatocyte damage, lipid disturbance, oxidative stress and elevated lipopolysaccharide levels were confirmed. Taurine treatment alleviated hepatocyte damage and oxidative stress. TPs treatment improved lipid metabolism and increased hepatic antioxidant activity. The therapeutic effects of taurine + TPs treatment on hepatocyte damage, lipid disturbance, and oxidative stress were superior to those of taurine and TPs treatment, respectively. Taurine, TPs and their combination all decreased serum lipopolysaccharide levels in NASH rats, but the combination of the compounds caused these levels to decrease more significantly than taurine or TPs treatment alone. CONCLUSION: Taurine combined with TPs treatment could relieve NASH by alleviating hepatocyte damage, decreasing oxidative stress and improving lipid metabolism and gut flora disturbance partly. Taurine and TPs combination may act as a new effective medicine for treating NASH patients.


Asunto(s)
Antioxidantes/farmacología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Polifenoles/farmacología , Taurina/farmacología , , Animales , Hígado/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
18.
Oncol Res ; 2017 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-28800793

RESUMEN

Histidine triad nucleotide binding protein 2 (HINT2) belongs to a HIT superfamily. It is ubiquitous and can be detectedin all forms of life. Recently, this protein has been reported to function as a tumor suppressor in some kind of cancers. However, little is known about its precise role in non-small cell lung cancer (NSCLC). In the present study, we explored the effects of HINT2 on the invasive potential of NSCLC cells. Our results showed that the expression of HINT2 was reduced in NSCLC tissues and cell lines. Up-regulation of HINT2 inhibited NSCLC cell invasion in vitro and tumor metastasis in vivo. We also found that the inhibitory effect of HINT2 on NSCLC cell invasion was associated with reduction of COX-2 expression. In addition, our further investigation on the underlying mechanisms demonstrated that up-regulation of HINT2 inhibited NSCLC cell invasion via COX-2/PGE2-mediated activation of ß-catenin signaling. In conclusion, we presented evidence that HINT2 exerted a tumor-suppression effect on the progression of NSCLC. Thus, HINT2 might be a promising molecular target for treatment of NSCLC.

19.
World J Gastroenterol ; 22(26): 6036-48, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27468195

RESUMEN

AIM: To evaluate the efficacy of umbilical cord-derived mesenchymal stem cells (UC-MSCs) transplantation in the treatment of liver fibrosis. METHODS: Cultured human UC-MSCs were isolated and transfused into rats with liver fibrosis induced by dimethylnitrosamine (DMN). The effects of UC-MSCs transfusion on liver fibrosis were then evaluated by histopathology; serum interleukin (IL)-4 and IL-10 levels were also measured. Furthermore, Kupffer cells (KCs) in fibrotic livers were isolated and cultured to analyze their phenotype. Moreover, UC-MSCs were co-cultured with KCs in vitro to assess the effects of UC-MSCs on KCs' phenotype, and IL-4 and IL-10 levels were measured in cell culture supernatants. Finally, UC-MSCs and KCs were cultured in the presence of IL-4 antibodies to block the effects of this cytokine, followed by phenotypical analysis of KCs. RESULTS: UC-MSCs transfused into rats were recruited by the injured liver and alleviated liver fibrosis, increasing serum IL-4 and IL-10 levels. Interestingly, UC-MSCs promoted mobilization of KCs not only in fibrotic livers, but also in vitro. Co-culture of UC-MSCs with KCs resulted in increased production of IL-4 and IL-10. The addition of IL-4 antibodies into the co-culture system resulted in decreased KC mobilization. CONCLUSION: UC-MSCs could increase IL-4 and promote mobilization of KCs both in vitro and in vivo, subsequently alleviating the liver fibrosis induced by DMN.


Asunto(s)
Interleucina-10/inmunología , Interleucina-4/inmunología , Macrófagos del Hígado/inmunología , Cirrosis Hepática/terapia , Hígado/inmunología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Cordón Umbilical/citología , Animales , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Dimetilnitrosamina/toxicidad , Modelos Animales de Enfermedad , Humanos , Técnicas In Vitro , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/inmunología , Cirrosis Hepática/patología , Ratas , Ratas Sprague-Dawley
20.
Oncotarget ; 7(26): 40508-40517, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27244892

RESUMEN

OBJECTIVE: To explore the expression feature and biological functions of TREM-1 on tumor-associated macrophages (TAMs) in lung cancer. RESULTS: The levels of TREM-1 on tissue-infiltrating monocytes/macrophage from tumor nest were significantly lower than those from nonturmor tissue or peripheral blood samples. Clinical analysis indicated that the levels of TREM-1-related TAMs were significantly decreased during cancer stages progression. The tumor-bearing mouse model further confirmed that the expression of TREM-1 on TAMs was significantly decreased with tumor growth. In addition, we found the activation of TREM-1 could significantly enhance the secretion of IL-1ß by TAM in vitro. Furthermore, T-bet but not Eomes was found to be the key transcription factor for the TREM-1 expression on monocytes/macrophage. METHODS: A total of 40 patients with non-small cell lung cancer (NSCLC) were enrolled in this study. The expression characteristics of TREM-1 in blood and tissue-infiltrating monocytes/macrophage were examined by flow cytometry analysis. After the treatment of TREM-1 antibody, which is an agonist of TREM-1, cytokines secreted by TAM were then analyzed. In LLC-tumor bearing mouse model, we further investigated the dynamic expression feature of TREM-1 on macrophage with tumor growth. Moreover, we explored the transcription factor for regulating TREM-1 expression on monocyes/macrophage with wildtype, T-bet Ko or Eomes Ko mice. CONCLUSION: The levels of TREM-1 were remarkably decreased during tumor progression. The low expression level of TREM-1 might be a characteristic for TAMs in lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Interleucina-1beta/metabolismo , Neoplasias Pulmonares/metabolismo , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Anciano , Animales , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Citocinas/metabolismo , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/diagnóstico , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Monocitos/citología , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA